US8435749B2 - Anti-CDH3 antibodies labeled with radioisotope label and uses thereof - Google Patents

Anti-CDH3 antibodies labeled with radioisotope label and uses thereof Download PDF

Info

Publication number
US8435749B2
US8435749B2 US13/001,869 US200913001869A US8435749B2 US 8435749 B2 US8435749 B2 US 8435749B2 US 200913001869 A US200913001869 A US 200913001869A US 8435749 B2 US8435749 B2 US 8435749B2
Authority
US
United States
Prior art keywords
antibody
cdh3
fragment
antibodies
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active, expires
Application number
US13/001,869
Other languages
English (en)
Other versions
US20120128584A1 (en
Inventor
Akira Togashi
Masakazu Katsu
Megumi Takayanagi
Hiroki Yoshioka
Pohsing Ng
Yasuhiro Shiba
Yusuke Nakamura
Keigo Endo
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Oncotherapy Science Inc
Original Assignee
Oncotherapy Science Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US13/001,869 priority Critical patent/US8435749B2/en
Application filed by Oncotherapy Science Inc filed Critical Oncotherapy Science Inc
Assigned to ONCOTHERAPY SCIENCE, INC. reassignment ONCOTHERAPY SCIENCE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NOSAN CORPORATION
Assigned to ONCOTHERAPY SCIENCE, INC. reassignment ONCOTHERAPY SCIENCE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KATSU, MASAKAZU, SHIBA, YASUHIRO, TAKAYANAGI, MEGUMI, YOSHIOKA, HIROKI, TOGASHI, AKIRA
Assigned to NOSAN CORPORATION reassignment NOSAN CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NG, POHSING
Assigned to NATIONAL UNIVERSITY CORPORATION GUNMA UNIVERSITY reassignment NATIONAL UNIVERSITY CORPORATION GUNMA UNIVERSITY ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ENDO, KEIGO
Assigned to THE UNIVERSITY OF TOKYO reassignment THE UNIVERSITY OF TOKYO ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NAKAMURA, YUSUKE
Assigned to ONCOTHERAPY SCIENCE, INC. reassignment ONCOTHERAPY SCIENCE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NATIONAL UNIVERSITY CORPORATION GUNMA UNIVERSITY
Assigned to ONCOTHERAPY SCIENCE, INC. reassignment ONCOTHERAPY SCIENCE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THE UNIVERSITY OF TOKYO
Publication of US20120128584A1 publication Critical patent/US20120128584A1/en
Publication of US8435749B2 publication Critical patent/US8435749B2/en
Application granted granted Critical
Active legal-status Critical Current
Adjusted expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • A61K51/1027Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody against receptors, cell-surface antigens or cell-surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/58Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances
    • G01N33/60Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving labelled substances involving radioactive labelled substances
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants

Definitions

  • the present invention relates to methods for damaging cancer cells using anti-CDH3 antibodies conjugated with radioisotope label, or to compositions for this purpose.
  • Cadherins are cell-cell adhesion glycoproteins that form calcium-dependent intercellular junctions and play an essential role in morphogenesis and in the development and maintenance of adult tissues and organs (Conacci-Sorrell M, et al., J Clin Invest, 109:987-91, (2002)).
  • the cell expression of specific cadherins results in homophilic interactions that are critical in the process of cell sorting and tissue stratification (Nose A, et al., Cell, 54:993-1001, (1988), Steinberg M S, et al., Proc Natl Acad Sci USA, 91:206-9, (1994) and Takeichi M. Science, 251:1451-5, (1991)).
  • the cadherin family is subdivided into various subfamilies, including the classical E-, P-, and N-cadherins, each demonstrating a specific tissue distribution (Takeichi M. Development, 102:639-55 (1988)).
  • E-cadherin is expressed in all epithelial tissues
  • P-cadherin CDH3
  • CDH3 is only restricted to the basal or lower layers of stratified epithelia, including prostate and skin, and also to the breast myoepithelial cells (Takeichi M. J Cell Biol 103:2649-58, (1986) and Shimoyama Y, et al., Cancer Res, 49:2128-33 (1989)).
  • Monoclonal antibodies against cancer-specific molecules have been proved to be useful in cancer treatment (Harris, M. (2004). Lancet Oncol, 5, 292-302.).
  • humanized or chimeric antibodies such as trastuzumab (Baselga, J. (2001). Oncology, 61, Suppl 2 14-21.), rituximab (Maloney, D. G., et al. (1997). Blood, 90, 2188-2195.) and bevacizumab (Ferrara, N., et al. (2004).
  • the present invention provides monoclonal antibodies against CDH3, which specifically recognize the polypeptide having the amino acid sequence as shown in SEQ ID NO: 3.
  • In vivo tumor-binding activity of this antibody was demonstrated using a fluorescent in vivo imaging system with near-infrared fluorescence in addition to the conventional method with radionuclides.
  • the present invention provides evidence of significant antitumor effect in xenograft mice bearing several cancer cell lines, wherein the mice are treated with a single or twice administration of 90Y-labeled anti-CDH3 monoclonal antibodies (clone#6) and chimeric thereof (ch-#6).
  • the present invention relates to the following:
  • the antibody or a fragment thereof which specifically recognizes a polypeptide having an amino acid sequence as shown in SEQ ID NO: 3.
  • the antibody or a fragment of the present invention comprises an H (heavy) chain V (variable) region comprising a complementarity determining region (CDR) having the amino acid sequences shown in SEQ ID NOs: 13, 14 and 15 or a CDR or functionally equivalent thereto (i.e., binds to the same antigenic determinant) and an L (light) chain V region comprising a CDR having the amino acid sequences shown in SEQ ID NOs: 16, 17 and 18 a CDR functionally equivalent thereto (i.e., binds to the same antigenic determinant), and which is capable of specifically binding to a CDH3 protein or a partial peptide thereof.
  • the antibody is selected from the group consisting of a mouse antibody, a chimeric antibody, a humanized antibody, a human antibody, an antibody fragment, and single-chain antibody
  • the antibody or fragment thereof of the invention can be labeled with a radioisotope label.
  • the radioisotope label is selected from the group consisting of 90yttrium (90Y), 125iodine (125I) and 111 indium (111In).
  • a composition, for treating or preventing cancer comprising a pharmaceutically effective amount of the antibody or fragment thereof of the invention.
  • the cancer is a pancreatic, lung, colon, prostate, breast, gastric or liver cancer.
  • a method for treating or preventing cancer in a subject comprising administering to said subject a pharmaceutically effective amount of the antibody or fragment thereof of the invention.
  • the cancer is a pancreatic, lung, colon, prostate, breast, gastric or liver cancer.
  • a method for diagnosis or prognosis of a disease that is associated with CDH3 or of a predisposition to develop the disease in a subject comprising
  • the cancer is a pancreatic, lung, colon, prostate, breast, gastric or liver cancer cell.
  • kits for diagnosis or prognosis of a disease associated with CDH3, comprising the antibody or fragment of the present invention.
  • the cancer is a pancreatic, lung, colon, prostate, breast, gastric or liver cancer.
  • FIG. 1 shows the results of CDH3 expression in cell lines.
  • Upper panels show flow cytometory analysis using each antibody.
  • KLM-1 and H358 are CDH3 positive cancer cell lines.
  • MIAPaCa-2 is a negative control cell line.
  • Anti-erbB2 is used as positive control. All of disclosed anti-CDH3 antibody clones can specifically recognize the cells expressing CDH3.
  • Lower panels are photographs depicting the results of Semiquantitative RT-PCR analysis for the CDH3 gene in cancer cell lines.
  • FIG. 2A shows in vivo fluorescence imaging of tumor-bearing mice after injection of each Alexa 647-labeled antibody clone. Fluorescence-labeled antibody clones are administered intravenously. All fluorescence images were acquired at 3, 24, 48 and 72 hours after injection. Fluorescence signal from Alexa647 was pseudo-colored which means that red is high density.
  • FIG. 2B show biodistributions of 111 In-labeled anti-CDH3 antibodies in EBC1 human lung cancer xenografted mice respectively (B: clone1).
  • FIG. 2C-D show biodistributions of 111 In-labeled anti-CDH3 antibodies in EBC1 human lung cancer xenografted mice respectively (C: clone3, D: clone4).
  • FIG. 2E-F show biodistributions of 111 In-labeled anti-CDH3 antibodies in EBC1 human lung cancer xenografted mice respectively (E: clone5, F: clone6).
  • FIG. 3A-B shows biodistribution of 111 In-labeled clone#6 antibody or control antibody.
  • the grafted tumors (A: EBC-1, B: SW948), liver, kidney, intestines, stomach, spleen, pancreas, lung, heart, muscle and bones are isolated at 48 hours after injection, and the radio-activities were measured.
  • Black box shows the anti-CDH3 antibody's accumulation to the each tissue.
  • FIG. 3C-D shows biodistribution of 111 In-labeled clone#6 antibody or control antibody.
  • the grafted tumors (C: KLM-1, D: H133), liver, kidney, intestines, stomach, spleen, pancreas, lung, heart, muscle and bones are isolated at 48 hours after injection, and the radio-activities were measured.
  • Black box shows the anti-CDH3 antibody's accumulation to the each tissue.
  • FIG. 3E shows biodistribution of 111 In-labeled anti-CDH3 chimeric or mouse antibodies #6, or normal human IgG or mouse IgG1 as control.
  • the grafted tumors (H133), liver, kidney, intestines, stomach, spleen, pancreas, lung, heart, muscle and bones are isolated at 48 hours after injection, and the radio-activities were measured.
  • Black box shows the anti-CDH3 chimeric antibody's accumulation to the each tissue.
  • White box shows the anti-CDH3 mouse antibody's accumulation to the each tissue.
  • FIG. 4 shows the effect of 90 Y-labeled anti-CDH3 antibody (clone#6) on tumor growth.
  • FIG. 4A shows that single administration of 90 Y-labeled anti-CDH3 antibody (clone#6) suppresses the growth of EBC-1 cells grafted in nude mice.
  • FIG. 4B shows HE-staining of the tumor tissue after the injection. As shown in the right panels, the tumor tissue substitutes for fiber tissue stained by the eosin (red color).
  • FIG. 5A shows that single administration of 90 Y-labeled anti-CDH3 antibody (clone#6) suppresses the growth of SW948 cells grafted in nude mice.
  • FIG. 5B shows the photograph of tumors in appearance. After the 90 Y-labeled anti-CDH3 antibody injection, the tumor apparently reduces.
  • FIG. 6 shows that single administration of 90 Y-labeled anti-CDH3 antibody (clone#6) suppresses the growth of KLM-1 cells grafted in nude mice.
  • FIG. 7 shows that both single and twice administration of 90 Y-labeled anti-CDH3 antibody (clone#6) suppresses the growth of SW948 cells grafted in nude mice.
  • FIG. 8 shows that single administration of 90 Y-labeled anti-CDH3 chimeric antibody (ch-#6) suppresses the growth of KLM-1 cells grafted in nude mice.
  • CHX and SCN represent p-SCN-Bn-CHX-A′′-DTPA and p-SCN-Bn-DTPA, respectively.
  • Antibodies are conjugated to these bifunctional chelators for radiolabeling with 90 Y.
  • the present invention relates to anti-CDH3 antibody and compositions and their use in treating or preventing a cancer.
  • the antibody is labeled with a radioisotope label.
  • antibody as used herein is intended to include immunoglobulins and fragments thereof which are specifically reactive to the designated protein or peptide thereof.
  • An antibody can include human antibodies, primatized antibodies, chimeric antibodies, bispecific antibodies, humanized antibodies, antibodies fused to other proteins or radiolabels, and antibody fragments.
  • an antibody herein is used in the broadest sense and specifically covers intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments so long as they exhibit the desired biological activity.
  • An “antibody” indicates all classes (e.g. IgA, IgD, IgE, IgG and IgM).
  • antibody fragments is a portion of an intact antibody, generally comprises the antigen binding or variable region of the intact antibody. Accordingly, in the present invention, antibody fragment may comprise antigen binding portion of the intact antibody.
  • the term “antigen-binding portion” of an antibody refers to one or more immunological active fragments of an antibody that retain the ability to specifically bind to an antigen (e.g., CDH3). It has been shown that the antigen-binding function of an antibody can be performed by fragments of a full-length antibody. Examples of antibody fragments include Fab, Fab′, F(ab′)2, and Fv fragments; linear antibodies; and single chain antibody molecules.
  • an antibody fragment binds with the same antigen that is recognized by the intact antibody.
  • antibody fragment also includes a synthetic or a genetically engineered polypeptide that binds to a specific antigen, such as polypeptides consisting of the light chain variable region, “Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker (“scFv proteins”), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.
  • scFv proteins recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker
  • variable regions substantially corresponding the sequences of the antibodies of the invention can vary from the referred-to sequence and still specifically recognize the same antigenic determinant.
  • This variation from the sequence may be stated in terms of a percentage of identical amino acids within the sequence. In some embodiments, this percentage is from about 90% to about 100%, e.g., 95, 96, 97, 98, 99, or 100%
  • nucleic refers to two or more sequences or subsequences that are the same or have a specified percentage of amino acid residues or nucleotides that are the same (i.e., 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or higher identity over a specified region, when compared and aligned for maximum correspondence over a comparison window or designated region) as measured using a BLAST or BLAST 2.0 sequence comparison algorithms with default parameters described below, or by manual alignment and visual inspection (see, e.g., NCBI web site at ncbi.nlm.nih.gov/BLAST/ or the like).
  • sequences are then said to be “substantially identical.”
  • This definition also refers to, or may be applied to, the compliment of a test sequence.
  • the definition also includes sequences that have deletions and/or additions, as well as those that have substitutions.
  • the preferred algorithms can account for gaps and the like.
  • identity exists over a region that is at least about 25 amino acids in length, or more preferably over a region that is 50-100 amino acids in length.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated.
  • sequence algorithm program parameters Preferably, default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • a “comparison window,” as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are well-known in the art. Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman, Adv. Appl. Math. 2:482 (1981), by the homology alignment algorithm of Needleman & Wunsch, J. Mol. Biol.
  • BLAST and BLAST 2.0 are used, with the parameters described herein, to determine percent sequence identity for the nucleic acids and proteins of the invention.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • the subject invention uses antibodies to CDH3. These antibodies will be provided by known methods.
  • Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen (e.g., SEQ ID NO: 3) and an adjuvant. It may be useful to conjugate the relevant antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOC12, or R′N ⁇ C ⁇ NR, where R′ and R are different alkyl groups.
  • a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyan
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g. 100 mcg or 5 mcg of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites.
  • the animals are boosted with 1 ⁇ 5 to 1/10 the original amount of peptide or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites.
  • Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus.
  • the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent.
  • Conjugates also can be made in recombinant cell culture as protein fusions. Also, aggregating agents such as alum are suitably used to enhance the immune response.
  • Monoclonal antibodies are obtained from a population of substantially homogeneous antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts.
  • the modifier “monoclonal” indicates the character of the antibody as not being a mixture of discrete antibodies.
  • the monoclonal antibodies may be made using the hybridoma method first described by Kohler et al., Nature, 256: 495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567).
  • a mouse or other appropriate host animal such as a hamster
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Manassas, Va., USA.
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133: 300 1 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the binding affinity of the monoclonal antibody can, for example, be determined by the Scatchard analysis of Munson et al., Anal. Biochem., 107: 220 (1980).
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of murine antibodies).
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese Hamster Ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • Another method of generating specific antibodies, or antibody fragments, reactive against a CDH3 is to screen expression libraries encoding immunoglobulin genes, or portions thereof, expressed in bacteria with a CDH3 protein or peptide.
  • complete Fab fragments, VH regions and Fv regions can be expressed in bacteria using phage expression libraries. See for example, Ward et al., Nature 341: 544-546 (1989); Huse et al., Science 246: 1275-1281 (1989); and McCafferty et al., Nature 348: 552-554 (1990).
  • Screening such libraries with, for example, a CDH3 peptide can identify immunoglobulin fragments reactive with CDH3.
  • the SCID-hu mouse available from Genpharm) can be used to produce antibodies or fragments thereof.
  • antibodies or antibody fragments can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348: 552-554 (1990). Clackson et al., Nature, 352: 624-628 (1991) and Marks et al., J. MoL BioL, 222: 581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc. Natl. Acad. ScL USA, 81: 6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • antibodies that bind to extracellular domain of CDH3 protein having the amino acid sequence of SEQ ID NO: 3 can be used for the present invention. Accordingly, as long as antibodies recognize SEQ ID NO: 3, such antibodies may be used for a method, or composition of the present invention. In preferred embodiments, antibodies recognizing the amino acid sequence of SEQ ID NO: 3 may bind to cells expressing CDH3 comprising at least transmembrane and extracellular domains thereof.
  • the present invention also provides an antibody suitable for treating or diagnosing CDH3 associated disease.
  • the antibody defined with following properties is preferable for such use in purpose.
  • VH and VL domains of antibodies of the present invention each include three CDRs designated as CDR1, CDR2, and CDR3, separated by framework regions.
  • Amino acid sequences of the CDRs are not particularly limited so long as the antibody can specifically bind to CDH3.
  • Examples of preferred CDR amino acid sequences include, but are not limited to:
  • VH CDR1 SYWIH
  • VH CDR2 EIDPSDNYTYYNQNFKG
  • VH CDR3 SGYGNLFVY
  • VL CDR1 SATSSVTYMY
  • VL CDR2 RTSNLAS
  • SEQ ID NO: 17 VL CDR3: QHYHIYPRT
  • VH corresponds to the amino acid sequence of SEQ ID NO: 11
  • VL corresponds to the amino acid sequence of SEQ ID NO: 12.
  • the monoclonal antibodies and binding fragments thereof may be characterized as:
  • RNA sequences coding for the chimeric and CDR-grafted products may be prepared, for example, by using the polymerase chain reaction (PCR) to synthesize the variable region from RNA of antibody-producing cells (see, for example, Larrick et al., “Methods: a Companion to Methods in Enzymology”, vol. 2: page 106 (1991); Courtenay-Luck, “Genetic Manipulation of Monoclonal Antibodies” in Monoclonal Antibodies: Production, Engineering and Clinical Application; Ritter et al.
  • PCR polymerase chain reaction
  • DNA sequences coding for the chimeric and CDR-grafted products may be synthesised completely or in part using oligonucleotide synthesis techniques. Site-directed mutagenesis and polymerase chain reaction techniques may be used as appropriate. For example, oligonucleotide directed synthesis as described by Jones et al., (1986) Nature.; 321:522-5 may be used.
  • oligonucleotide directed mutagenesis of a pre-existing variable region as, for example, described by Verhoeyen et al., (1988) Science.; 239:1534-6 or Riechmann et al., (supra) may be used.
  • enzymatic filling in of gapped oligonucleotides using T4 DNA polymerase as, for example, described by Queen et al., (1989) Proc Natl Acad Sci USA.; 86:10029-33; PCT Publication WO 90/07861 may be used.
  • Any suitable host cell/vector system may be used for expression of the DNA sequences coding for the CDR-grafted heavy and light chains.
  • Bacterial, e.g., E. coli , and other microbial systems may be used, in particular for expression of antibody fragments such as FAb and (Fab′) 2 fragments, and especially Fv fragments and single-chain antibody fragments, e.g., single-chain Fvs.
  • Eucaryotic, e.g., mammalian, host cell expression systems may be used, in particular, for production of larger CDR-grafted antibody products, including complete antibody molecules.
  • Suitable mammalian host cells include CHO cells and myeloma or hybridoma cell lines.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321: 522-525 (1986); Reichmann et al., Nature, 332: 323-327 (1988); Verhoeyen et al., Science, 239: 1534-1536 (1988)), by substituting hypervariable region sequences for the corresponding sequences of a human antibody.
  • humanized antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567) wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species.
  • humanized antibodies are typically human antibodies in which some hypervariable region residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • variable domains both light and heavy
  • the choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity.
  • the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the rodent is then accepted as the human framework region (FR) for the humanized antibody (Suns et al., J. Immunol., 151: 2296 (1993); Chothia et al., J. Mol. Biol, 196: 901 (1987)).
  • Another method uses a particular framework region derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains.
  • the same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89: 4285 (1992); Presta et al., J. Immunol., 151: 2623 (1993)).
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen, is achieved.
  • the hypervariable region residues are directly and most substantially involved in influencing antigen binding.
  • human antibodies can be generated.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • JH antibody heavy-chain joining region
  • transfer of the human germ-line immunoglobulin gene array in such germ line mutant mice will result in the production of human antibodies upon antigen challenge. See, e.g., Jakobovits et al., Proc. Mad. Acad. Sci.
  • phage display technology can be used to produce human antibodies and antibody fragments in vitro, from immunoglobulin variable (V) domain gene repertoires from unimmunized donors.
  • V domain genes are cloned in-frame into either a major or minor coat protein gene of a filamentous bacteriophage, such as M13 or fd, and displayed as functional antibody fragments on the surface of the phage particle. Because the filamentous particle contains a single-stranded DNA copy of the phage genome, selections based on the functional properties of the antibody also result in selection of the gene encoding the antibody exhibiting those properties.
  • the phage mimics some of the properties of the B cell.
  • Phage display can be performed in a variety of formats; for their review see, e.g., Johnson, Kevin S, and Chiswell, David J., Current Opinion in Structural Biology 3: 564-571 (1993).
  • V-gene segments can be used for phage display.
  • Human antibodies may also be generated by in vitro activated B cells (see U.S. Pat. Nos. 5,567,610 and 5,229,275). A preferred means of generating human antibodies using SCID mice is disclosed in commonly-owned, co-pending applications.
  • antibody fragments Traditionally, these fragments were derived via proteolytic digestion of intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods 24: 107-117 (1992) and Brennan et al., Science, 229: 81 (1985)). However, these fragments can now be produced directly by recombinant host cells. For example, the antibody fragments can be isolated from the antibody phage libraries discussed above. Alternatively, Fab′-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab′) 2 fragments (Carter et al., Bio/Technology 10: 163-167 (1992)).
  • F(ab′) 2 fragments can be isolated directly from recombinant host cell culture.
  • the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; U.S. Pat. No. 5,571,894; and U.S. Pat. No. 5,587,458.
  • the antibody fragment may also be a “linear antibody”, e.g., as described in U.S. Pat. No. 5,641,870. Such linear antibody fragments may be monospecific or bispecific.
  • Bispecific antibodies are antibodies that have binding specificities for at least two different epitopes.
  • an anti-cancer cell marker binding arm may be combined with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc receptors for IgG (FcyR), such as FcyRI (CD64), FcyRII (CD32) and FcyRIH(CD 16) so as to focus cellular defense mechanisms to the cancer cell.
  • Bispecific antibodies may also be used to localize cytotoxic agents to the cancer cell. These antibodies possess a cancer cell marker-binding arm and an arm which binds the cytotoxic agent (e.g.
  • Bispecific antibodies can be prepared as full length antibodies or antibody fragments (e.g. F(ab) 2 bispecific antibodies).
  • bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305: 537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBO J., 10: 3655-3659 (1991).
  • antibody variable domains with the desired binding specificities are fused to immunoglobulin constant domain sequences.
  • the fusion preferably is with an immunoglobulin heavy chain constant domain, comprising at least part of the hinge, CH2, and CH3 regions. It is preferred to have the first heavy-chain constant region (CHI) containing the site necessary for light chain binding, present in at least one of the fusions.
  • DNAs encoding the immunoglobulin heavy chain fusions and, if desired, the immunoglobulin light chain are inserted into separate expression vectors, and are co-transfected into a suitable host organism.
  • the bispecific antibodies are composed of a hybrid immunoglobulin heavy chain with a first binding specificity in one arm, and a hybrid immunoglobulin heavy chain-light chain pair (providing a second binding specificity) in the other arm. It was found that this asymmetric structure facilitates the separation of the desired bispecific compound from unwanted immunoglobulin chain combinations, as the presence of an immunoglobulin light chain in only one half of the bispecific molecule provides for a facile way of separation. This approach is disclosed in WO 94/04690. For further details of generating bispecific antibodies see, for example, Suresh et al., Methods in Enzymology, 121: 210 (1986).
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • the preferred interface comprises at least a part of the CH3 domain of an antibody constant domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan).
  • Compensatory “cavities” of identical or similar size to the large side chains) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • Bispecific antibodies include cross-linked or “heteroconjugate” antibodies.
  • one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin.
  • Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089).
  • Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.
  • bispecific antibodies can be prepared using chemical linkage.
  • Brennan et al., Science, 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab′) 2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation.
  • the Fab′ fragments generated are then converted to thionitrobenzoate (TNB) derivatives.
  • One of the Fab′-TNB derivatives is then reconverted to the Fab′-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab′-TNB derivative to form the bispecific antibody.
  • the bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
  • bispecific antibodies have been produced using leucine zippers (Kostelny et al., J Immunol. 148 (5): 1547-1553 (1992)).
  • the leucine zipper peptides from the Fos and Jun proteins were linked to the Fab′ portions of two different antibodies by gene fusion.
  • the antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers.
  • the fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites.
  • VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites.
  • sFv single-chain Fv
  • Antibodies with more than two valencies are contemplated.
  • trispecific antibodies can be prepared. Tutt et al. J; Immunol. 147: 60 (1991).
  • the antibodies used in the methods or included in the articles of manufacture herein are optionally conjugated to a cytotoxic or therapeutic agent.
  • a therapeutic agent includes any chemotherapeutic agent which is useful in the treatment of cancer.
  • chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylmelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolmelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide,
  • paclitaxel (TAXOLO, Bristol-Myers Squibb Oncology, Princeton, N.J.) and doxetaxel (TAXOTEW, Rh6ne-Poulenc Rorer, Antony, France); chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as cisplatin and carboplatin; vinblastine; platinum; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylornithine (DMFO); retinoic acid; esperamicins; capecitabine; and pharmaceutically acceptable salts, acids or derivatives of any of the
  • anti-hormonal agents that act to regulate or inhibit hormone action on tumors
  • anti-estrogens including for example tamoxifen, raloxifene, aromatase inhibiting 4 (5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically acceptable salts, acids or derivatives of any of the above.
  • Conjugates of an antibody and one or more small molecule toxins such as a calicheamicin, a maytansine (U.S. Pat. No. 5,208,020), a trichothecene, and CC 1065 are also contemplated herein.
  • the antibodies are conjugated to one or more maytansine molecules (e.g. about 1 to about 10 maytansine molecules per antibodies molecule).
  • Maytansine may, for example, be converted to May SS-Me which may be reduced to May-SH 3 and reacted with modified antibodies (Chari et al. Cancer Research 52: 127-131 (1992)) to generate a maytansinoid-antibody conjugate.
  • the antibody may be conjugated to one or more calicheamicin molecules.
  • the calicheamicin family of antibiotics is capable of producing double stranded DNA breaks at sub-picomolar concentrations.
  • Structural analogues of calicheamicin which may be used include, but are not limited to gamma1I, alpha2I, alpha3I, N-acetyl-gammalI, PSAG and OI1 (Hinman et al. Cancer Research 53: 3336-3342 (1993) and Lode et al, Cancer Research 58: 2925-2928 (1998)).
  • Enzymatically active toxins and fragments thereof which can be used include diphtheria A chain, nonbinding active fragments of diphtheria toxin, exotoxin A chain (from Pseudomonas aeruginosa ), ricin A chain, abrin A chain, modeccin A chain, alpha sarcin, Aleurites fordii proteins, dianthin proteins, Phytolaca americana proteins (PAPI, PAPII, and PAP-S), momordica charantia inhibitor, curcin, crotin, sapaonaria officinalis inhibitor, gelonin, mitogellin, restrictocin, phenomycin, enomycin and the trichothecenes. See, for example, WO 93/21232 published Oct. 28, 1993.
  • the present invention further contemplates antibody conjugated with a variety of radioactive isotopes. Examples include 111 In, 211 At, 131 I, 125 I, 90 Y, 186 Re, 188 Re, 153 Sm, 212 Bi, 32 P and radioactive isotopes of Lu.
  • antibody of the present invention may be labeled with radio-nuclides just prior to use, or provided as radio-labeled antibody.
  • radio-nuclides and chemocytotoxic agents that can be coupled to tumor-specific antibodies by well-known techniques and delivered to a site to specifically damaging tumor cells and tissue. (See, for example, U.S. Pat. No. 4,542,225 to W. A.
  • imaging and cytotoxic reagents that are suitable for use include 125 I, 123 I. 111 In (e.g., Sumerdon et al., 1990, Nucl. Med. Biol., 17:247-254), and 99m Tc; fluorescent labels such as fluorescein and rhodamine; chemiluminescent labels such as luciferin, and paramagnetic ions for use in magnetic resonance imaging (Lauffer et al., 1991, Magnetic Resonance in Medicine, 22:339-342).
  • Antibodies can be labeled with such reagents using protocols and techniques known and practiced in the art. See, for example, Wenzel and Meares, Radioimmunoimaging and Radioimmunotherapy, Elsevier, N.Y., 1983; Colcer et al., 1986, Meth. Enzymol., 121:802-816; and Monoclonal Antibodies for Cancer Detection and Therapy, Eds. Baldwin et al., Academic Press, 1985, pp. 303-316, for techniques relating to the radiolabeling of antibodies.
  • Yttrium-90 ( 90 Y) labeled monoclonal antibodies have been described for maximizing the dose delivered to the tumor or cancer cells and/or tissue, while limiting toxicity to normal tissues (e.g., Goodwin and Meares, 1997, Cancer Supplement, 80:2675-2680).
  • Other cytotoxic radionuclides including, but not limited to, Iodine-131 ( 131 I) and Rhenium-186 can also be used for labeling monoclonal antibodies of the present invention.
  • Yttrium-90 may be suitable for radioimmunotherapy, since Yttrium-90 ( 90 Y) provides advantages over Iodine-131 ( 131 I) because it delivers higher beta energy (2.3 MeV vs 0.61 MeV) to the tumor and has path length of 5 to 10 mm resulting in the improved ability to kill both targeted and neighboring cells, an advantage particularly in bulky or poorly vascularized tumor.
  • the detectable/detecting label used is selected according to the imaging modality to be used.
  • radioactive labels such as Indium-111 ( 111 In), Technetium-99m ( 99m Tc), or Iodine 131 ( 131 I)
  • SPECT single photon emission computed tomography
  • positron-emitting labels such as Fluorine-19 can be used in positron emission tomography (PET).
  • Paramagnetic ions such as Gadlinium(III) or Manganese(II) can be used in magnetic resonance imaging (MRI).
  • the monoclonal antibodies can also be labeled with radio-opaque labels for the visualization of cancer cells after injection, for example, by X-ray, CATscan, or MRI.
  • radio-opaque labels for the visualization of cancer cells after injection, for example, by X-ray, CATscan, or MRI.
  • CDH3 relating disease (e.g. cancers)
  • localization of the label within the cancers permits the determination of the spread of the disease.
  • the amount of label that is present and detectable within the cancers expressing CDH3, for example, allows the determination of the presence or absence of cancer or tumor in the subject to be diagnosed.
  • Conjugates of the antibody and cytotoxic agent may be made using a variety of bifunctional protein coupling agents such as N-succinimidyl-3-(2-pyriylditliol) propionate (SPDP), succinimidyl-4-(N-maleimidomethyl)cyclohexane-1-carboxylate, iminothiolane (IT), bifunctional derivatives of imidoesters (such as dimethyl adipimidate HCL), active esters (such as disuccinimidyl suberate), aldehydes (such as glutareldehyde), bis-azido compounds (such as bis(p-azidobenzoyl) hexanediamine), bis-diazonium derivatives (such as bis-(p-diazoniumbenzoyl)-ethylenediamine), diisocyanates (such as tolyene 2,6-diisocyanate), and bis-active fluorine compounds (such as 1,5-diflu
  • a ricin immunotoxin can be prepared as described in Vitetta et al. Science 238: 1098 (1987).
  • Carbon-14-labeled 1-isothiocyanatobenzyl-3-methyldiethylene triaminepentaacetic acid (MX-DTPA) is an exemplary chelating agent for conjugation of radionucleotide to the antibody (See WO94/11026).
  • the linker may be a “cleavable linker” facilitating release of the cytotoxic drug in the cell.
  • an acid-labile linker, peptidase-sensitive linker, dimethyl linker or disulfide-containing linker (Charm et al. Cancer Research 52: 127-131 (1992)) may be used.
  • a fusion protein comprising the antibody and cytotoxic agent may be made, e.g. by recombinant techniques or peptide synthesis.
  • the antibody may be conjugated to a “receptor” (such streptavidin) for utilization in tumor pretargeting wherein the antibody-receptor conjugate is administered to the patient, followed by removal of unbound conjugate from the circulation using a clearing agent and then administration of a “ligand” (e.g. avidin) which is conjugated to a cytotoxic agent (e.g. a radionucleotide).
  • a “receptor” such streptavidin
  • a ligand e.g. avidin
  • cytotoxic agent e.g. a radionucleotide
  • the antibodies of the present invention may also be conjugated with a prodrug activating enzyme which converts a prodrug (e.g. a peptidyl chemotherapeutic agent, see WO81/01145) to an active anti-cancer drug (See, for example, WO 88/07378 and U.S. Pat. No. 4,975,278).
  • a prodrug activating enzyme which converts a prodrug (e.g. a peptidyl chemotherapeutic agent, see WO81/01145) to an active anti-cancer drug (See, for example, WO 88/07378 and U.S. Pat. No. 4,975,278).
  • the enzyme component of such conjugates includes any enzyme capable of acting on a prodrug in such a way so as to covert it into its more active, cytotoxic form.
  • Enzymes that are useful in the method of this invention include, but are not limited to, alkaline phosphatase useful for converting phosphate-containing prodrugs into free drugs; arylsulfatase useful for converting sulfate-containing prodrugs into free drugs; cytosine deaminase useful for converting non-toxic5-fluorocytosine into the anti-cancer drug, fluorouracil; proteases, such as serratia protease, thermolysin, subtilisin, carboxypeptidases and cathepsins (such as cathepsins B and L), that are useful for converting peptide-containing prodrugs into free drugs; D-alanylcarboxypeptidases, useful for converting prodrugs that contain D-amino acid substituents; carbohydratecleaving enzymes such as 13-galactosidase and neuraminidase useful for converting glycosylated prodrugs into free drugs; 13-lactama
  • antibodies with enzymatic activity can be used to convert the prodrugs of the invention into free active drugs (see, e.g., Massey, Nature 328: 457-458 (1987)).
  • Antibody-abzyme conjugates can be prepared as described herein for delivery of the abzyme to a tumor cell population.
  • the enzymes of this invention can be covalently bound to the antibody by techniques well known in the art such as the use of the heterobifunctional crosslinking reagents discussed above.
  • fusion proteins comprising at least the antigen binding region of an antibody of the invention linked to at least a functionally active portion of an enzyme of the invention can be constructed using recombinant DNA techniques well known in the art (see, e.g., Neuberger et al., Nature, 312: 604-608 (1984)).
  • the antibody may be linked to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • nonproteinaceous polymers e.g., polyethylene glycol, polypropylene glycol, polyoxyalkylenes, or copolymers of polyethylene glycol and polypropylene glycol.
  • Liposomes containing the antibody are prepared by methods known in the art, such as described in Epstein et al., Proc. Natl. Acad. Sci. USA, 82: 3688 (1985); Hwang et al., Proc. Natl. Acad. Sci. USA, 77: 4030 (1980); U.S. Pat. Nos. 4,485,045 and 4,544,545; and WO97/38731 published Oct. 23, 1997. Liposomes with enhanced circulation time are disclosed in U.S. Pat. No. 5,013,556.
  • Particularly useful liposomes can be generated by the reverse phase evaporation method with a lipid composition comprising phosphatidylcholine, cholesterol and PEG derivatized phosphatidylethanolamine (PEG-PE). Liposomes are extruded through filters of defined pore size to yield liposomes with the desired diameter.
  • Fab′ fragments of an antibody of the present invention can be conjugated to the liposomes as described in Martin et al. J; Biol. Chem. 257: 286-288 (1982) via a disulfide interchange reaction.
  • a chemotherapeutic agent is optionally contained within the liposome (See Gabizon et al. A National Cancer Inst. 81 (19) 1484 (1989)).
  • Amino acid sequence modifications of antibodies described herein are contemplated. For example, it may be desirable to improve the binding affinity and/or other biological properties of the antibody.
  • Amino acid sequence variants of the antibody are prepared by introducing appropriate nucleotide changes into the antibody encoding nucleic acid, or by peptide synthesis. Such modifications include, for example, deletions from, and/or insertions into and/or substitutions of, residues within the amino acid sequences of the antibody. Any combination of deletion, insertion, and substitution is made to arrive at the final construct, provided that the final construct possesses the desired characteristics.
  • the amino acid changes also may alter post-translational processes of the antibody, such as changing the number or position of glycosylation sites.
  • a useful method for identification of certain residues or regions of the antibody that are preferred locations for mutagenesis is called “alanine scanning mutagenesis” as described by Cunningham and Wells Science, 244: 1081-1085 (1989).
  • a residue or group of target residues are identified (e.g., charged residues such as arg, asp, his, lys, and glu) and replaced by a neutral or negatively charged amino acid (most preferably alanine or polyalanine) to affect the interaction of the amino acids with antigen.
  • Those amino acid locations demonstrating functional sensitivity to the substitutions then are refined by introducing further or other variants at, or for, the sites of substitution.
  • the site for introducing an amino acid sequence variation is predetermined, the nature of the mutation per se need not be predetermined. For example, to analyze the performance of a mutation at a given site, ala scanning or random mutagenesis is conducted at the target codon or region and the expressed antibody variants are screened for the desired activity
  • Amino acid sequence insertions include amino- and/or carboxyl-terminal fusions ranging in length from one residue to polypeptides containing a hundred or more residues, as well as intrasequence insertions of single or multiple amino acid residues.
  • terminal insertions include an antibody with an N-terminal methionyl residue or the antibody fused to a cytotoxic polypeptide.
  • Other insertional variants of the antibody molecule include the fusion to the N- or C-terminus of the antibody of an enzyme, or a polypeptide which increases the serum half-life of the antibody.
  • variants are an amino acid substitution variant. These variants have at least one amino acid residue in the antibody molecule replaced by different residue.
  • the sites of greatest interest for substitutional mutagenesis of antibody include the hypervariable regions, but FR alterations are also contemplated.
  • Substantial modifications in the biological properties of the antibody are accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the polypeptide backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, or (c) the bulk of the side chain.
  • hydrophobic norleucine, met, ala, val, leu, ile
  • Non-conservative substitutions will entail exchanging a member of one of these classes for another class.
  • cysteine residue not involved in maintaining the proper conformation of the antibody also may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking.
  • cysteine bonds may be added to the antibody to improve its stability (particularly where the antibody is a fragment such as an Fv fragment).
  • a particularly preferred type of substitutional variant involves substituting one or more hypervariable region residues of a parent antibody (e.g. a humanized or human antibody).
  • a parent antibody e.g. a humanized or human antibody
  • the resulting variants selected for further development will have improved biological properties relative to the parent antibody from which they are generated.
  • a convenient way for generating such substitutional variants is affinity maturation using phage display. Briefly, several hypervariable region sites (e.g. 6-7 sites) are mutated to generate all possible amino substitutions at each site.
  • the antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M13 packaged within each particle. The phage-displayed variants are then screened for their biological activity (e.g. binding affinity) as herein disclosed.
  • alanine scanning mutagenesis can be performed to identify hypervariable region residues contributing significantly to antigen binding.
  • Another type of amino acid variant of the antibody alters the original glycosylation pattern of the antibody. By altering is meant deleting one or more carbohydrate moieties found in the antibody, and/or adding one or more glycosylation sites that are not present in the antibody.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • O-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly seine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • the alteration may also be made by the addition of, or substitution by, one or more seine or threonine residues to the sequence of the original antibody (for O-linked glycosylation sites).
  • Nucleic acid molecules encoding amino acid sequence variants of the antibody are prepared by a variety of methods known in the art. These methods include, but are not limited to, isolation from a natural source (in the case of naturally occurring amino acid sequence variants) or preparation by oligonucleotide-mediated (or site-directed) mutagenesis, PCR mutagenesis, and cassette mutagenesis of an earlier prepared variant or a non-variant version of the antibody.
  • ADCC antigen-dependent cell-mediated cyotoxicity
  • CDC complement dependent cytotoxicity
  • This may be achieved by introducing one or more amino acid substitutions in an Fc region of an antibody.
  • cysteine residue(s) may be introduced in the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC). See Caron et al., J. Exp Med. 176: 1191-1195 (1992) and Shopes, B. J limmunol 148: 2918-2922 (1992).
  • Homodimeric antibodies with enhanced anti-tumor activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al. Cancer Research 53: 2560-2565 (1993).
  • an antibody can be engineered which has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities (See Stevenson et al. Anti-Cancer Drug Design 3: 219-230 (1989)).
  • a salvage receptor binding epitope refers to an epitope of the Fc region of an IgG molecule (e.g., IgG1, IgG2, IgG3, or IgG4) that is responsible for increasing the in vivo serum half-life of the IgG molecule.
  • An anti-CDH3 antibody of the present invention may be used as a marker for diagnosing a disease that is associated with CDH3.
  • the present invention provides methods for diagnosing a disease that is associated with CDH3 or a predisposition for developing a disease that is associated with CDH3 in a subject by detecting the CDH3 protein with an anti-CDH3 antibody of the present invention in the subject.
  • the methods comprise the steps of:
  • a disease that is associated with CDH3 is pancreatic, lung, colon, prostate, breast, gastric or liver cancer.
  • the anti-CDH3 antibody of the present invention may be used for detecting or imaging a cancer in a living body. More specifically, the present invention provides methods of detecting or imaging a cancer which comprise the steps of:
  • cancer cells or tissues may be detected in a patient.
  • the present invention provides methods for detecting a cancer, in which CDH3 is expressed in tumor tissue in a patient, comprising: administering an antibody or antibody fragment to the subject allowing the antibody or antibody fragment specifically binds to CDH3 in the cells or tissue; visualizing the antibody bound in the cells or tissue; and comparing levels of antibody bound in the cells or tissue to a normal control cells or tissue, wherein an increase in the level of antibody bound to the patient cells or tissue relative to the normal control cells or tissue is indicative of a cancer in the patient.
  • the antibody in order to trace the antibody administered into a living body, the antibody may be labeled with detectable molecules.
  • detectable molecules For example, the behavior of antibodies labeled with a fluorescent substance, luminescent substance, or radioisotope can be traced in vivo. Methods for labeling an antibody with such molecules are well known in the art.
  • Antibodies labeled with a fluorescent substance or a luminescent substance can be observed, for example, using an endoscope or a laparoscope.
  • the localization of an antibody can be imaged by tracing the radioactivity of the radioisotope.
  • the localization of an anti-CDH3 antibody in vivo demonstrates the presence of cancer cells.
  • an anti-CDH3 antibody of the present invention may be used for in vivo imaging.
  • an anti-CDH3 antibody of the present invention labeled for detection may be used in order to visualize CDH3 protein in a living body.
  • the behavior of antibodies labeled with a fluorescent substance, luminescent substance, or radioisotope can be traced in vivo.
  • Antibodies labeled with a fluorescent substance or a luminescent substance can be observed using bioimaging system.
  • the localization of an antibody can be imaged by immunoscintigraphy.
  • Another embodiment of the present invention provides diagnostics, diagnostic methods and imaging methods for cancers and tumors expressing CDH3 using the monoclonal antibodies or binding fragments of the present invention.
  • the diagnostic uses of the antibody of the present invention embrace primary tumors and cancers, as well as metastases.
  • cancers expressing CDH3 may be selected from the group consisting of pancreatic, lung, colon, prostate, breast, gastric or liver cancers.
  • a diagnostic method comprises administering, introducing, or infusing the monoclonal antibody or their binding fragments as described herein, with or without conjugation to a detectable moiety, such as a radioisotope.
  • a detectable moiety such as a radioisotope.
  • the antibody or binding fragment binds to the tumor or cancer cells, after which the location of the bound antibodies or fragments is detected.
  • imaging instrumentation may be used to identify the location of the agent within the body.
  • a second detectable reagent may be administered, which locates the bound antibodies or fragments so that they can be suitable detected.
  • mcg. microgram
  • mcg. preferably about 50-500 mcg, more preferably about 100-300 mcg, more preferably about 200-300 mcg of purified antibody
  • applicable doses for humans include about 100-200 mcg/kg body weight, or 350-700 mg/m2 of body-surface area.
  • an intermediate result for examining the condition of a subject may be provided.
  • Such intermediate result may be combined with additional information to assist a doctor, nurse, or other practitioner to determine that a subject suffers from a disease that is associated with CDH3.
  • the present invention may be used to detect cancerous cells in a subject-derived tissue, and provide a doctor with useful information to determine that the subject suffers from pancreatic, lung, colon, prostate, breast, gastric or liver cancers.
  • the differentially expressed CDH3 gene also allows for the course of treatments for a disease that is associated with CDH3 e.g., pancreatic cancer cells (PaC cells), lung cancer cells (LuC cells), colon cancer cells (CC cells), prostate cancer cells (e.g., PrC cells), breast cancer cells (BC cells), gastric cancer cells (GC cells), or liver cancer cells (LiC cells) to be monitored and assessed.
  • a disease e.g., pancreatic cancer cells (PaC cells), lung cancer cells (LuC cells), colon cancer cells (CC cells), prostate cancer cells (e.g., PrC cells), breast cancer cells (BC cells), gastric cancer cells (GC cells), or liver cancer cells (LiC cells)
  • an intermediate result for monitoring the course of treatment of PaC, LuC, CC, PrC, BC, GC, or LiC may be provided.
  • CDH3 gene or protein encoded thereby is useful prognostic marker for monitoring clinical outcome of PaC, LuC, CC, PrC, BC, GC, or LiC.
  • the present invention may be used to detect cancerous cells in a subject-derived tissue, and provide a doctor with useful information to assess the course of treatment of PaC, LuC, CC, PrC, BC, GC, or LiC.
  • a test cell population is provided from a subject undergoing treatment for PaC, LuC, CC, PrC, BC, GC, or LiC.
  • test cell populations are obtained from the subject at various time points, before, during, and/or after treatment. Expression of the CDH3 gene in the test cell population is then determined and compared to expression of the same genes in a reference cell population which includes cells whose PaC, LuC, CC, PrC, BC, GC, or LiC state is known. In the context of the present invention, the reference cells should not have been exposed to the treatment of interest.
  • the biological sample should be derived from a subject undergoing treatment for pancreatic, lung, colon, prostate, breast, gastric or liver cancers.
  • multiple test biological samples are obtained from the subject at various time points before, during or after the treatment.
  • the reference cell population contains non-PaC cells, non-LuC cells, non-CC cells, non-PrC cells, non-BC cells, non-GC cells, or non-LiC cells
  • a similarity in the expression of the CDH3 gene in the test cell population and the reference cell population indicates that the treatment of interest is efficacious.
  • a difference in the expression of the CDH3 gene in the test cell population and a normal control reference cell population indicates a less favorable clinical outcome or prognosis.
  • the reference cell population contains PaC cells, LuC cells, CC cells, PrC cells, BC cells, GC cells, or LiC cells
  • a difference between the expression of the CDH3 gene in the test cell population and the reference cell population indicates that the treatment of interest is efficacious
  • a similarity in the expression of the CDH3 gene in the test population and an PaC, LuC, CC, PrC, BC, GC, or LiC control reference cell population indicates a less favorable clinical outcome or prognosis.
  • the expression level of the CDH3 gene determined in a biological sample from a subject obtained after treatment can be compared to the expression level of the CDH3 gene determined in a biological sample from a subject obtained prior to treatment onset (i.e., pre-treatment levels).
  • a decrease in the expression level in a post-treatment sample indicates that the treatment of interest is efficacious while an increase or maintenance in the expression level in the post-treatment sample indicates a less favorable clinical outcome or prognosis.
  • the term “efficacious” indicates that the treatment leads to a reduction in the expression of CDH3 gene or a decrease in size, prevalence, or metastatic potential of PaC, LuC, CC, PrC, BC, GC, or LiC in a subject.
  • the term “efficacious” means that the treatment retards or prevents a lung cancer and/or an esophageal tumor from forming or retards, prevents, or alleviates a symptom of clinical PaC, LuC, CC, PrC, BC, GC, or LiC.
  • Assessment of pancreatic, lung, colon, prostate, breast, gastric or liver tumors can be made using standard clinical protocols.
  • efficaciousness can be determined in association with any known method for diagnosing or treating PaC, LuC, CC, PrC, BC, GC, or LiC.
  • PaC, LuC, CC, PrC, BC, GC, or LiC can be diagnosed, for example, histopathologically or alternatively by identifying symptomatic anomalies, e.g., weight loss, loss of appetite, abdominal pain, back pain, anorexia, nausea, vomiting and generalized malaise, weakness, and jaundice.
  • the present invention also provides methods for assessing the prognosis of a subject with a disease that is associated with CDH3, e.g., PaC, LuC, CC, PrC, BC, GC, or LiC, such methods including the step of comparing the expression of the CDH3 gene in a test cell population to the expression of the CDH3 gene in a reference cell population from patients over a spectrum of disease stages.
  • a disease that is associated with CDH3, e.g., PaC, LuC, CC, PrC, BC, GC, or LiC
  • the prognosis of the subject can be assessed.
  • an intermediate result for assessing the prognosis of a subject with PaC, LuC, CC, PrC, BC, GC, or LiC may be provided.
  • Such intermediate result may be combined with additional information to assist a doctor, nurse, or other practitioner to determine that a subject suffers from lung cancer or esophageal cancer.
  • the present invention may be used to detect cancerous cells in a subject-derived tissue, and provide a doctor with useful information to assess the prognosis of a subject with PaC, LuC, CC, PrC, BC, GC, or LiC.
  • an increase in the expression of the CDH3 gene in a test sample as compared to a normal control sample indicates a less favorable prognosis.
  • a similarity in the expression of the CDH3 gene, in a test sample as compared to normal control sample indicates a more favorable prognosis for the subject.
  • the present invention provides a kit for diagnosis or prognosis of a disease associated with CDH3.
  • the kit includes a reagent for detecting the CDH3 protein.
  • Suitable reagents for detecting the CDH3 protein include an antibody to the CDH3 protein.
  • the antibody may be labeled with detectable molecules.
  • the antibody may be labeled with fluorescent substance, luminescent substance, or radioisotope.
  • kits may include positive and negative control reagents, and a secondary antibody for detecting an antibody against the CDH3 protein.
  • tissue samples obtained from subjects with good prognosis or poor prognosis may serve as useful control reagents.
  • a kit of the present invention may further include other materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes, and package inserts (e.g., written, tape, CD-ROM, etc.) with instructions for use. These reagents and such may be retained in a container with a label. Suitable containers include bottles, vials, and test tubes. The containers may be formed from a variety of materials, such as glass or plastic.
  • the present invention further provides a kit for use in detecting, imaging or treating a cancer within a subject to be diagnosed comprising the antibody against the CDH3 protein.
  • the antibody of the present invention may be labeled with a radioisotope.
  • the kit of the present invention may contain an antibody recognizing CDH3 modified with chelating agent and radioactive substance. MX-DOPA is preferable chelating agent for modifying the antibody.
  • indium-111 ( 111 In) can be used as a tracer for bioimaging.
  • antibody may be labeled with beta nuclides e.g. Yttrium-90 ( 90 Y).
  • indium-111 ( 111 In) or Yttrium-90 ( 90 Y) may also be provided as salt or solution thereof.
  • Suitable salt of indium-111 ( 111 In) or Yttrium-90 ( 90 Y) is chloride.
  • a disease associated with CDH3 is pancreatic, lung, colon, prostate, breast, gastric or liver cancers.
  • the cancer includes but is not limited to a pancreatic, lung, colon, prostate, breast, gastric or liver cancer cell.
  • the method for treating and/or preventing the cancer in a subject according to the present invention comprises administering to a subject in need thereof the antibody or the fragment described above.
  • subject herein refers to a subject who has suffered from the cancer including but is not limited to a pancreatic, lung, colon, prostate, breast, gastric or liver cancer cell.
  • the subject in the present invention may be animals including mammals and avian animals.
  • mammals may include humans, mice, rats, monkeys, rabbits, and dogs.
  • the antibody or fragment thereof described herein can specifically bind to CDH3 protein, so when the antibody or fragment thereof is administered to a subject, it binds to CDH3 protein in the subject and the growth of the cells expressing CDH3 may be suppressed.
  • the antibody or fragment thereof may be conjugated with a therapeutic moiety and administered to a subject, it is delivered to a region that expresses CDH3 protein (i.e. suffered region) in a subject and the therapeutic moiety can be selectively delivered to the suffered region and acted thereon.
  • Such therapeutic moiety may be any therapeutics that are known or will be developed for having a therapeutic efficacy on the cancer and includes, but not limited to, a radioisotope label and chemotherapeutic agent.
  • a radioisotope label which can be used as therapeutics can be selected depending on a variety of elements including beta-ray energy and its emission efficiency, the presence or absence of gamma-ray emitted, its energy and emission efficiency, physical half-life, and labeling procedure.
  • the radioisotope label based on yttrium (such as 90 Y) and iodine (such as 125 I and 131 I) may be used.
  • a chemotherapeutic agent may be any agent that is known or will be developed for treating the cancer and includes, but not limited to, methotrexate, taxol, mercaptopurine, thioguanine, cisplatin, carboplatin, mitomycin, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin, vinblastine, vincristine, vinorelbine, paclitaxel, and docetaxel.
  • the antibody or fragment thereof described herein can selectively bind to CDH3 protein and not bind to a normal cell, so side effect which is caused by the antibody or fragment thereof, or radioisotope or chemotherapeutic agent can be effectively avoided and therefore the therapeutic potency may be high.
  • the antibody or fragment thereof described herein can be administered to a subject at effective doses to treat or prevent the CDH3-associated disease.
  • An effective dose refers to that amount of an antibody or a fragment thereof sufficient to result in a healthful benefit in the treated subject.
  • Formulations and methods of administration that can be employed when the pharmaceutical composition contains an antibody of the present invention are described below.
  • a cocktail of different monoclonal antibodies such as a mixture of the specific monoclonal antibodies described herein or their binding fragments, may be administered, if necessary or desired, to alleviate cancers.
  • a mixture of monoclonal antibodies, or binding fragments thereof, in a cocktail to target several antigens, or different epitopes, on cancer cells is an advantageous approach, particularly to prevent evasion of tumor cells and/or cancer cells due to downregulation of one of the antigens.
  • compositions for use in accordance with the present invention can be formulated in conventional manner using one or more pharmaceutically acceptable carriers or excipients.
  • the antibodies or fragments thereof can be formulated for parenteral administration (i.e., intravenous or intramuscular) by injection, via, for example, bolus injection or continuous infusion.
  • parenteral administration i.e., intravenous or intramuscular
  • Formulations for injection can be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • the compositions can take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the antibody can be in lyophilized powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • Toxicity and therapeutic efficacy of the antibody or fragment, or the therapeutic moiety conjugated thereto can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD/ED.
  • Antibodies or therapeutic moieties that exhibit large therapeutic indices are preferred. While antibodies or moieties that exhibit toxic side effects can be used, care should be taken to design a delivery system that targets such antibodies or moieties to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • the data obtained from the cell culture assays and animal studies can be used in formulating a range of dosages for use in humans.
  • the dosage of such antibodies lies preferably within a range of circulating plasma concentrations that include the ED50 with little or no toxicity.
  • the dosage can vary within this range depending upon the dosage form employed, the route of administration utilized and types and amounts of the therapeutic moiety conjugated.
  • the effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating plasma concentration range that includes the IC50 (i.e., the concentration of the test antibody that achieves a half-maximal inhibition of symptoms) as determined in cell culture.
  • IC50 i.e., the concentration of the test antibody that achieves a half-maximal inhibition of symptoms
  • levels in plasma can be measured, for example, by high performance liquid chromatography.
  • the pharmaceutical composition of the present invention is administered in an amount such that the antibody according to the present invention is administered to the subject in a day in an amount of about 3 to about 15 mcg per kg body weight of subject, and preferably of about 10 to about 15 mcg per kg body weight of subject.
  • the administration interval and times can be selected in consideration of the condition and age of the subject, administration route, and response to the pharmaceutical composition.
  • the pharmaceutical composition can be administered to the subject one to 5 times, preferably 1 times a day for 5 to 10 days.
  • the administrative dose for a single adult is 0.1 mCi/kg to 1.0 mCi/kg, preferably 0.1 mCi/kg to 0.5 mCi/kg, and more preferably 0.4 mCi/kg at once.
  • the pharmaceutical composition can be administered systemically or locally. It is preferably administered in a targeting delivery manner so as to deliver the active component to an affected site.
  • the methods and compositions of the present invention are used for the treatment or prevention of the cancer together with one or a combination of chemotherapeutic agents including, but not limited to, methotrexate, taxol, mercaptopurine, thioguanine, cisplatin, carboplatin, mitomycin, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin, vinblastine, vincristine, vinorelbine, paclitaxel, and docetaxel.
  • chemotherapeutic agents including, but not limited to, methotrexate, taxol, mercaptopurine, thioguanine, cisplatin, carboplatin, mitomycin, bleomycin, doxorubicin, idarubicin, daunorubicin, dactinomycin, vinblastine, vincristine, vinorelbine, paclitaxel, and docetaxel.
  • any radiation therapy protocol can be used depending upon the type of the cancer to be treated.
  • X-ray radiation can be administered.
  • Gamma ray emitting radioisotopes such as radioactive isotopes of radium, cobalt, and other elements may also be administered to expose tissues.
  • chemotherapy or radiation therapy is administered, preferably at least an hour, five hours, 12 hours, a day, a week, a month, and more preferably several months (e.g., up to three months) subsequent to using the methods and compositions containing the antibody of the present invention.
  • the chemotherapy or radiation therapy administered prior to, concurrently with, or subsequent to the treatment using the methods and compositions according to the present invention can be administered by any method known in the art.
  • the present invention also provides the use of the antibody of the present invention in manufacturing a pharmaceutical composition for treating or preventing a disease associated with CDH3.
  • the present invention further provides a use of radio-labeled antibody of the present invention for manufacturing a pharmaceutical composition for treating or preventing a cancer.
  • the present invention further provides the antibody of the present invention for use in treating or preventing a disease associated with CDH3.
  • the radio-labeled antibody of the present invention for use in radioimmunotherapy for cancer is also provided.
  • the present invention further provides a method or process for manufacturing a pharmaceutical composition for treating or preventing a disease associated with CDH3, wherein the method or process comprises step for formulating a pharmaceutically or physiologically acceptable carrier with the antibody of the present invention as active ingredients.
  • the present invention further provides a method or process for manufacturing a pharmaceutical composition for treating or preventing a cancer, wherein the method or process comprises step for formulating a pharmaceutically or physiologically acceptable carrier with the radio-labeled antibody of the present invention as active ingredients.
  • the present invention also provides a method or process for manufacturing a pharmaceutical composition for treating or preventing a disease associated with CDH3, wherein the method or process comprises step for admixing an active ingredient with a pharmaceutically or physiologically acceptable carrier, wherein the active ingredient is the antibody of the present invention.
  • the present invention further provides a method or process for manufacturing a pharmaceutical composition for treating or preventing a cancer, wherein the method or process comprises step for admixing the radio-labeled antibody of the present invention with a pharmaceutically or physiologically acceptable carrier.
  • the present invention provides the antibody of the present invention for use in bioimaging or immunoscintigraphy for cancer within a subject to be diagnosed.
  • the present invention provides use of the antibody of the present invention for manufacturing a diagnostic agent for bioimaging or immunoscintigraphy for cancer within a subject.
  • the present invention further provides a method or process for manufacturing a diagnostic agent for bioimaging or immunoscintigraphy for cancer within a subject, wherein the method or process comprises step for admixing the antibody of the present invention with a pharmaceutically or physiologically acceptable carrier.
  • CDH3 gene encoded extracellular domain (SEQ ID NO: 3) was amplified from cDNA pool derived from cancer cells. The product was cloned into the pcDNA3.1 (Invitrogen, CA). To produce CDH3-specific antibody, mice were immunized subcutaneously with the domain expression vector (17.5 mcg/injection) every two weeks for a month. After the confirmation of the titer of antisera, spleenocytes were extracted from the mice and fused to myeloma cells to prepare hybridomas. We screened the hybridomas which can recognize native CDH3 antigen on the surface of the cancer cells.
  • hybridoma clone #6 produced antigen-specific antibody at high level, therefore this clone was selected to produce antibody for further experiments.
  • the hybridoma clone #6 was used to inject intraperitoneally into mice. The ascites was recovered after 2 to 3 weeks. Finally antibody was purified from the ascites using Protein A column (GE Healthcare, NJ).
  • EBC-1, H1373 and H358 as non-small cell lung cancer line
  • KLM-1 and MIAPaCa-2 as pancreatic cancer line
  • SW948 colorectal cancer line. All cell lines were obtained from American Type Culture Collection (Manassasm, Va.), and were maintained in EMEM (EBC-1, MIAPaCa-2), RPMI (H358, KLM-1, H1373) and L-15 (SW948) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin at 37 degrees C. in a humidified atmosphere of 5% CO 2 .
  • FBS fetal bovine serum
  • H358, KLM-1 and MIAPaCa-2 were maintained in each recommended culture media until early-confluency.
  • the cells were subsequently dissociated with Cell Dissociation Solution (SIGMA, MO) to avoid denaturing of cell surface antigen.
  • Cells were suspended with PBS (1 ⁇ 107 cells/mL) and incubated with test antibody (50 mcg/mL) for 1 h at 4 degrees C. After washing twice with PBS, cells were mixed with 20 mcg/mL of Alexa Fluor488 goat anti-mouse IgG (H+L) (Invitrogen, CA) containing 7AAD (2.5 mcg/mL) and incubated for 30 min at 4 degrees C. in dark. Binding mode and specificity of test antibody was evaluated by FACS Calibur (Becton Dickinson, NJ) according to manufacturer's instruction.
  • antibody-chelator complexes were purified using Biospin Column 6 (Bio-Rad, Tokyo, Japan). 111 InCl 3 (Nihon Medi-Physics, Hyogo, Japan) and 90 YCl3 (QSA Global, Brauschweig, Germany) were respectively pre-incubated with 0.25 M acetic acid (pH 5.5) for 5 minutes at room temperature in parallel.
  • the antibody-chelator complexes were incubated with the preincubated 111 InCl 3 and 90 YCl 3 solution respectively for 1 hour at 37 degrees C.
  • Labeled antibody was purified using Biospin Column 6 according to manufacture's instructions. It was confirmed that degradation of these antibodies was not observed during labeling processes.
  • mice Animal care and treatment was performed in accordance with the guidelines of animal use and animal committee of the Gunma University. 100 mcL of EBC1, SW948, KLM-1 and H1373 cell suspension (1 ⁇ 10 7 cells) were inoculated subcutaneously into the right flank of female 3- to 5-week-old nude mice (Charles River Laboratories Japan Inc. Yokohama, Japan). These mice were kept for several weeks to develop the tumors. The established tumors were isolated from tumor-bearing mice and dissected into cubic tissue fragments 2 mm on a side. These fragments were transplanted serially into nude mice. After the transplantation, these mice were kept until the average tumor volume reached 100-600 mm3.
  • mice bearing established human lung cancer tumor EBC-1 were randomly selected and assigned to two groups. The mice were injected with Alexa647-labeled and 111 In-labeled (0.5 mCi/mg) antibodies intravenously. For fluorescence-based study, the biodistribution of Alexa647-labeled antibodies was evaluated with IVIS 200 bioimaging system (Caliper Life Sciences, MA) at 48 hours after treatment. For radioisotope-based study, the grafted tumors were isolated, along with blood, liver, kidney, intestines, stomach, spleen, pancreas, lung, heart, muscle and bones respectively at 3, 24, 48 and 72 hours after injection.
  • 111 In-labeled clone 6 was analyzed at 48 hours using nude mice bearing established tumors (EBC-1, SW948, KLM-1 and H133) respectively.
  • Xenograft mice were randomly assigned to three different treatment groups.
  • 90 Y-labeled antibodies (4-10 mCi/mg) were prepared as described above.
  • the mice were injected intravenously with 90 Y-labeled clone 6 or 90 Y-labeled normal mouse IgG1 as control. Radioactivity of injected antibodies was adjusted to 100 mcCi per animal.
  • Body weight and tumor volume of the treated-xenograft mice were monitored for 4 weeks after 90 Y-labeled antibody injection.
  • the tumor volume (mm 3 ) was calculated using following formula: (the shortest diameter) 2 ⁇ (the longest diameter) ⁇ 0.5.
  • SW948 xenograft mice were randomly assigned to four different treatment groups. Antibodies were conjugated to p-SCN-Bn-DTPA for radiolabeling with 90 Y. 90 Y-labeled antibodies (4-10 mCi/mg) were prepared as described above. The mice were injected intravenously with 90 Y-labeled clone 6 or 90 Y-labeled normal IgG1 as control. Second injection was performed with 90 Y-labeled clone 6 after 14 days of the first injection. Radioactivity of injected antibodies was adjusted to 100 mcCi per animal. Body weight and tumor volume of the treated-xenograft mice were monitored for 7 weeks after first 90 Y-labeled antibody injection. The tumor volume (mm3) was calculated using following formula: (the shortest diameter) 2 ⁇ (the longest diameter) ⁇ 0.5.
  • Isolated tumors were embedded with Lab-Tek OCT compound (Sakura Finetek USA, CA) on dry-ice to prepare frozen tissue section.
  • the frozen sections were washed with distilled water to remove OCT compound, and treated with xylene for 5 min in twice.
  • the sections were rinsed with 100%, 90%, 80%, 70% and 50% ethanol for 10 sec, respectively.
  • Slide sections were immersed in Mayer's Hematoxilin (Muto Pure Chemicals, Japan) for 5 min, and washed excessive Hematoxilin with flowing water. Next, dehydration was performed using 50%, 70%, 80%, 90% and 100% ethanol after 1% Eosin Y (Muto Pure Chemicals, Japan) treatment.
  • the sections were treated with xylene for 5 min in thrice, serially enclosed by Malinol (DAIDO SANGYO, Japan).
  • RNAs were extracted from hybridoma Clone #6 using RNeasy Mini Kit (QIAGEN). cDNA was synthesized from the total RNA using SuperScript II Reverse Transcriptase (Invitrogen). The sequences of variable regions of monoclonal antibodies were amplified using NovaTaq DNA polymerase (Novagen) and Mouse Ig-Primer Set (Novagen).
  • B as C, G or T, D as A, G or T, H as A, C or T, I as inosine, K as G or T, M as A or C, R as A or G, S as C or G, V as A, C or G, W as A or T and Y as C or T.
  • variable region of light chain was amplified by PCR using primers of SEQ ID NO: 25 and 26.
  • the constant region of human kappa was amplified by PCR using primers of SEQ ID NO: 29 and 30.
  • Gene of light chain was amplified with these two PCR products containing common sequence in terminal by overlap extension PCR using primers of SEQ ID NO: 32 and 30.
  • the digested heavy chain SGV DNA contains the hCMV-MIE promoter-heavy chain-SV40 transcription unit, and the digested light chain SGV DNA contains the GS transcription unit and the hCMV-MIE, promoter-light chain expression cassette. Both purified fragments were ligated to construct the double gene expression vector. E. coli cells were transformed with the vector and then the vector was prepared. The vector expressing both H-chain and L-chains was transfected into 293T cells. The medium was exchanged with serum-free medium (DMEM; GIBCO, 11965-092), and the chimeric antibody contained in the culture supernatant was purified via Protein A column.
  • DMEM serum-free medium
  • CDH3 antigen was presented on the surface of H358 and KLM-1, but not on MIAPaCa-2 ( FIG. 1 ).
  • the peak uptake of the clone 6 into the tumor was 32.0+/ ⁇ 3.5% ID/g (EBC-1), 21.4+/ ⁇ 2.3% ID/g (SW948), 24.2+/ ⁇ 1.2% ID/g (KLM-1), 23.8+/ ⁇ 3.1% ID/g (H1373) respectively at 48 h after injection ( FIG. 3A-D ).
  • the peak uptake of the chimeric antibody of clone 6 named ch-#6 into the tumor was 38.0+/ ⁇ 3.2% ID/g (H1373) at 48 h after injection ( FIG. 3E ).
  • the uptake of control IgG never exceeded 10% ID/g in every mouse ( FIG. 3A-E ).
  • radiotherapy with 90 Y-labeled clone 6 and ch-#6 was performed using xenograft mice. All tumors were drastically decreased growth rate by radiation from yttrium-90 conjugated with the clone 6 ( FIGS. 4A , 5 A, 6 , 7 ) and ch-#6 ( FIG. 8 ). On the other hand, 90 Y-labeled control antibody showed no effect on tumor growth ( FIGS. 4A , 5 A, 6 , 7 , 8 ). Therefore, this therapeutic effect seemed to depend on the antigen-antibody reaction.
  • tumor cell number was drastically decreased and the marked fibrosis was observed by the treatment with 90 Y-labeled clone 6. It is notable that no fibrosis was observed in tumors treated with 90 Y-labeled control IgG ( FIG. 4B ).
  • SW948 colon cancer
  • FIG. 5 The transplanted tumors almost disappeared in each mouse after 21 days of injection of 90 Y-labeled clone 6 ( FIG. 5B ).
  • KLM-1 pancreatic cancer
  • CDH3 is attractive therapeutic target for these tumors and anti-CDH3 antibody is usefultherapeutic drug candidate for this type of treatment.
  • amino acid sequence of mouse Ig H-chain variable regions and L-chain variable regions were determined as follows:
  • H-chain variable region (except for the signal sequence): (SEQ ID NO: 11) QVQLQQPGAELVKPGTSVKLSCKSSGYTFTSWIHWVKQRPGHGLEWIGEI DPSDNYTYYNQNFKGKATLTIDKSSSTAYMQLNSLTSEDSAVFYCARSGY GNLFVYWGQGTLVTVSA; and Clone #6, L-chain variable region (except for the signal sequence): (SEQ ID NO: 12) QIVLTQSPAIMSSSPGEKVTMSCSATSSVTYMYWYQQKPGSSPKPWIFRT SNLASGVPTRFSGSGSGTSYSLTISSMEAEDAATYYCQHYHIYPRTFGGG TKL.
  • the CDR (complementarity determining region) sequences of the antibodies were determined by the Kabat definition as follows:
  • SYWIH SEQ ID NO: 13
  • EIDPSDNYTYYNQNFKG SEQ ID NO: 14
  • SGYGNLFVY SEQ ID NO: 15
  • SATSSVTYMY SEQ ID NO: 16
  • RTSNLAS SEQ ID NO: 17
  • QHYHIYPRT SEQ ID NO: 18
  • the present invention is based, at least in part, on the discovery that a cancer expressing CDH3 can be treated with radioisotope labeled anti-CDH3 antibody in vivo.
  • CDH3 was identified by the present inventors as a gene strongly expressed in pancreatic, lung, colon, prostate, breast, gastric or liver cancers.
  • treatment of a cancer for example, pancreatic, lung, colon, prostate, breast, gastric or liver cancer is conveniently carried out using anti-CDH3 antibodies labeled with radioisotope label.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Physics & Mathematics (AREA)
  • Public Health (AREA)
  • Microbiology (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Epidemiology (AREA)
  • Optics & Photonics (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Mycology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US13/001,869 2008-06-30 2009-06-30 Anti-CDH3 antibodies labeled with radioisotope label and uses thereof Active 2029-09-23 US8435749B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/001,869 US8435749B2 (en) 2008-06-30 2009-06-30 Anti-CDH3 antibodies labeled with radioisotope label and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US7698208P 2008-06-30 2008-06-30
US13/001,869 US8435749B2 (en) 2008-06-30 2009-06-30 Anti-CDH3 antibodies labeled with radioisotope label and uses thereof
PCT/JP2009/003009 WO2010001585A1 (fr) 2008-06-30 2009-06-30 Anticorps anti-cdh3 marqués par un marqueur radioisotopique, et leurs utilisations

Publications (2)

Publication Number Publication Date
US20120128584A1 US20120128584A1 (en) 2012-05-24
US8435749B2 true US8435749B2 (en) 2013-05-07

Family

ID=41465693

Family Applications (1)

Application Number Title Priority Date Filing Date
US13/001,869 Active 2029-09-23 US8435749B2 (en) 2008-06-30 2009-06-30 Anti-CDH3 antibodies labeled with radioisotope label and uses thereof

Country Status (10)

Country Link
US (1) US8435749B2 (fr)
EP (1) EP2313504B1 (fr)
JP (1) JP5593560B2 (fr)
KR (1) KR20110025215A (fr)
CN (1) CN102137929B (fr)
BR (1) BRPI0913656A2 (fr)
CA (1) CA2729403A1 (fr)
ES (1) ES2534437T3 (fr)
RU (1) RU2011103199A (fr)
WO (1) WO2010001585A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130034566A1 (en) * 2009-12-28 2013-02-07 Onco Therapy Science, Inc. Anti-cdh3 antibodies and uses thereof
US20160058883A1 (en) * 2012-04-04 2016-03-03 Perseus Proteomics Inc. Drug conjugate comprising anti-cdh3 (p-cadherin) antibody
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US20180193477A1 (en) * 2015-07-15 2018-07-12 Zymeworks Inc. Drug-conjugated bi-specific antigen-binding constructs

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL2535358T3 (pl) * 2010-02-10 2018-05-30 Fujifilm Ri Pharma Co., Ltd. Przeciwciało przeciwko kadherynie znakowane radioaktywnym metalem
WO2012057328A1 (fr) * 2010-10-29 2012-05-03 株式会社ペルセウスプロテオミクス Anticorps anti-cdh3 présentant une capacité élevée d'internalisation
EP3138581B1 (fr) 2011-03-17 2019-01-02 The University of Birmingham Immunothérapie redirigée
JP6377601B2 (ja) 2013-02-15 2018-08-22 株式会社ペルセウスプロテオミクス 抗cdh3ヒト化抗体、その薬剤コンジュゲート、及びそれらの使用
ES2868305T3 (es) * 2014-03-28 2021-10-21 Univ Washington Through Its Center For Commercialization Vacunas contra el cáncer de mama y de ovario
KR102546611B1 (ko) * 2014-04-08 2023-06-22 보스턴 파마슈티칼즈, 아이엔씨. Il-21에 특이적인 결합 분자 및 이들 용도
TN2017000173A1 (en) 2014-11-14 2018-10-19 Novartis Ag Antibody drug conjugates
EP4276116A3 (fr) * 2015-04-17 2024-01-17 Amgen Research (Munich) GmbH Constructions d'anticorps bispécifiques pour cdh3 et cd3
JP6974348B2 (ja) * 2016-01-09 2021-12-01 アーベル リミテッドArbele Limited がん処置のためのカドヘリン−17特異的抗体及び細胞傷害性細胞
JP7104274B2 (ja) * 2016-08-19 2022-07-21 Pdrファーマ株式会社 緩衝剤を含む組成物
CN108396063B (zh) * 2017-02-07 2021-11-02 复旦大学附属华山医院 Cdh23基因突变在垂体腺瘤分子诊断中的应用
CN109239758B (zh) * 2018-07-27 2022-08-12 国家海洋局南海环境监测中心(中国海监南海区检验鉴定中心) 一种生物样品中伽马能谱核素检测分析方法
KR20210105890A (ko) 2018-12-17 2021-08-27 레비토프 리미티드 트윈 면역 세포 인게이저
CN117285631A (zh) * 2023-11-24 2023-12-26 原子高科股份有限公司 用于放射性免疫治疗的Lu-177标记MUC1抗体

Citations (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002097395A2 (fr) 2001-05-31 2002-12-05 Chiron Corporation Utilisation de p-cadherine comme cible dans la therapie anticancereuse
WO2004001072A2 (fr) 2002-06-19 2003-12-31 Oncotherapy Science, Inc. Procede permettant de diagnostiquer des tumeurs colo-rectales
WO2004000020A1 (fr) 2002-06-24 2003-12-31 Bayer Cropscience Aktiengesellschaft Combinaisons de substances actives fongicides
WO2004007770A2 (fr) 2002-07-10 2004-01-22 Oncotherapy Science, Inc. Procede de diagnostic de tumeurs gastriques de type intestinal
WO2004024952A1 (fr) 2002-08-30 2004-03-25 Oncotherapy Science, Inc. Méthode de diagnostic de l'endométriose ovarienne
WO2004031413A2 (fr) 2002-09-30 2004-04-15 Oncotherapy Science, Inc. Technique de diagnostic de cancers bronchopulmonaires « non a petites cellules »
WO2004031410A2 (fr) 2002-09-30 2004-04-15 Oncotherapy Science, Inc. Methode permettant de diagnostiquer des seminomes du testicule
WO2004031412A2 (fr) 2002-09-30 2004-04-15 Oncotherapy Science, Inc. Methode de diagnostic du cancer du pancreas
WO2004110345A2 (fr) 2002-10-29 2004-12-23 Pharmacia Corporation Genes exprimes de maniere differenciee impliques dans le cancer, polypeptides codes par ces genes, et methodes d'utilisation des genes
WO2005000020A2 (fr) 2003-06-23 2005-01-06 Cognis Ip Management Gmbh Vecteurs d'alcool d'alkoxylate pour principes actifs pesticides
US20050214836A1 (en) 2002-08-30 2005-09-29 Oncotherapy Science, Inc. Method of diagnosing ovarian endometriosis
WO2005090572A2 (fr) 2004-03-24 2005-09-29 Oncotherapy Science, Inc. Compositions et procédés servant à traiter un cancer du pancréas
US20050260639A1 (en) 2002-09-30 2005-11-24 Oncotherapy Science, Inc. Method for diagnosing pancreatic cancer
WO2006000020A1 (fr) 2004-06-29 2006-01-05 European Nickel Plc Lixiviation amelioree de metaux de base
US20060024692A1 (en) 2002-09-30 2006-02-02 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
WO2006085684A2 (fr) 2005-02-10 2006-08-17 Oncotherapy Science, Inc. Methode de diagnostic du cancer de la vessie
US20060246077A1 (en) * 2001-12-28 2006-11-02 Menashe Bar-Eli Use of antibodies against the MUC18 antigen
WO2006114704A2 (fr) 2005-04-26 2006-11-02 Pfizer Inc. Anticorps de la p-cadherine
WO2007000020A1 (fr) 2005-06-29 2007-01-04 Compumedics Limited Ensemble capteur a pont conducteur
WO2007075672A2 (fr) 2005-12-23 2007-07-05 Lankenau Institute For Medical Research Marqueurs pour le pronostic du cancer
EP1830187A1 (fr) 2002-10-15 2007-09-05 Technion Research And Development Foundation, Ltd. Procédés et compositions pour moduler la croissance des cheveux via des modulateurs de P-cadhérine
WO2007102525A1 (fr) 2006-02-28 2007-09-13 Oncotherapy Science, Inc. Procédés permettant d'endommager des cellules au moyen des fonctions effectrices des anticorps anti-cdh3
NL1031818C2 (nl) 2006-05-15 2007-11-23 Pfizer P-Cadherineantilichamen.
WO2008000020A1 (fr) 2006-06-29 2008-01-03 Fermiscan Australia Pty Limited Détection de substance anormale dans un échantillon de kératine prélevé chez des sujets présentant une pathologie
WO2008102557A1 (fr) 2007-02-21 2008-08-28 Oncotherapy Science, Inc. Vaccins peptidiques pour les cancers exprimant des antigènes associés à une tumeur
WO2009025116A1 (fr) 2007-08-20 2009-02-26 Oncotherapy Science, Inc. Peptide cdh3 et agent médicinal le comprenant
US20100040641A1 (en) 2007-02-21 2010-02-18 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing tumor-associated antigens

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1994A (en) 1841-02-23 Manner of fastening and gombining ti-ie truss-frames of bblidges
US912069A (en) 1907-03-06 1909-02-09 Jesse T Burr Heater.
WO1981001145A1 (fr) 1979-10-18 1981-04-30 Univ Illinois Medicaments "pro-drugs" pouvant etre actives par des enzymes hydrolytiques
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4542225A (en) 1984-08-29 1985-09-17 Dana-Farber Cancer Institute, Inc. Acid-cleavable compound
GB8705477D0 (en) 1987-03-09 1987-04-15 Carlton Med Prod Drug delivery systems
US4975278A (en) 1988-02-26 1990-12-04 Bristol-Myers Company Antibody-enzyme conjugates in combination with prodrugs for the delivery of cytotoxic agents to tumor cells
ZA932522B (en) 1992-04-10 1993-12-20 Res Dev Foundation Immunotoxins directed against c-erbB-2(HER/neu) related surface antigens
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life

Patent Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002097395A2 (fr) 2001-05-31 2002-12-05 Chiron Corporation Utilisation de p-cadherine comme cible dans la therapie anticancereuse
US20060246077A1 (en) * 2001-12-28 2006-11-02 Menashe Bar-Eli Use of antibodies against the MUC18 antigen
WO2004001072A2 (fr) 2002-06-19 2003-12-31 Oncotherapy Science, Inc. Procede permettant de diagnostiquer des tumeurs colo-rectales
US20060199179A1 (en) 2002-06-19 2006-09-07 Oncotherapy Science, Inc. Method for diagnosis of colorectal tumors
WO2004000020A1 (fr) 2002-06-24 2003-12-31 Bayer Cropscience Aktiengesellschaft Combinaisons de substances actives fongicides
US20060105333A1 (en) 2002-07-10 2006-05-18 Oncotherapy Science, Inc. Method for diagnosis of intestinal-type gastric tumors
WO2004007770A2 (fr) 2002-07-10 2004-01-22 Oncotherapy Science, Inc. Procede de diagnostic de tumeurs gastriques de type intestinal
US20050214836A1 (en) 2002-08-30 2005-09-29 Oncotherapy Science, Inc. Method of diagnosing ovarian endometriosis
WO2004024952A1 (fr) 2002-08-30 2004-03-25 Oncotherapy Science, Inc. Méthode de diagnostic de l'endométriose ovarienne
US20060194199A1 (en) 2002-09-30 2006-08-31 Oncotherapy Science, Inc. Method for diagnosing testicular seminomas
US20050260639A1 (en) 2002-09-30 2005-11-24 Oncotherapy Science, Inc. Method for diagnosing pancreatic cancer
US20060024692A1 (en) 2002-09-30 2006-02-02 Oncotherapy Science, Inc. Method for diagnosing non-small cell lung cancers
WO2004031412A2 (fr) 2002-09-30 2004-04-15 Oncotherapy Science, Inc. Methode de diagnostic du cancer du pancreas
WO2004031410A2 (fr) 2002-09-30 2004-04-15 Oncotherapy Science, Inc. Methode permettant de diagnostiquer des seminomes du testicule
WO2004031413A2 (fr) 2002-09-30 2004-04-15 Oncotherapy Science, Inc. Technique de diagnostic de cancers bronchopulmonaires « non a petites cellules »
US20090162361A1 (en) 2002-09-30 2009-06-25 Oncotherapy Science, Inc. Method for diagnosing pancreatic cancer
EP1830187A1 (fr) 2002-10-15 2007-09-05 Technion Research And Development Foundation, Ltd. Procédés et compositions pour moduler la croissance des cheveux via des modulateurs de P-cadhérine
WO2004110345A2 (fr) 2002-10-29 2004-12-23 Pharmacia Corporation Genes exprimes de maniere differenciee impliques dans le cancer, polypeptides codes par ces genes, et methodes d'utilisation des genes
WO2005000020A2 (fr) 2003-06-23 2005-01-06 Cognis Ip Management Gmbh Vecteurs d'alcool d'alkoxylate pour principes actifs pesticides
WO2005090572A2 (fr) 2004-03-24 2005-09-29 Oncotherapy Science, Inc. Compositions et procédés servant à traiter un cancer du pancréas
WO2006000020A1 (fr) 2004-06-29 2006-01-05 European Nickel Plc Lixiviation amelioree de metaux de base
WO2006085684A2 (fr) 2005-02-10 2006-08-17 Oncotherapy Science, Inc. Methode de diagnostic du cancer de la vessie
US20090175844A1 (en) 2005-02-10 2009-07-09 Oncotherapy Science Inc. Method of diagnosing bladder cancer
US20120014996A1 (en) 2005-02-10 2012-01-19 Oncotherapy Science, Inc. Method of diagnosing bladder cancer
WO2006114704A2 (fr) 2005-04-26 2006-11-02 Pfizer Inc. Anticorps de la p-cadherine
WO2007000020A1 (fr) 2005-06-29 2007-01-04 Compumedics Limited Ensemble capteur a pont conducteur
WO2007075672A2 (fr) 2005-12-23 2007-07-05 Lankenau Institute For Medical Research Marqueurs pour le pronostic du cancer
WO2007102525A1 (fr) 2006-02-28 2007-09-13 Oncotherapy Science, Inc. Procédés permettant d'endommager des cellules au moyen des fonctions effectrices des anticorps anti-cdh3
US20090169572A1 (en) 2006-02-28 2009-07-02 Oncotherapy Science, Inc, Methods for damaging cells using effector functions of anti-cdh3 antibodies
NL1031818C2 (nl) 2006-05-15 2007-11-23 Pfizer P-Cadherineantilichamen.
WO2008000020A1 (fr) 2006-06-29 2008-01-03 Fermiscan Australia Pty Limited Détection de substance anormale dans un échantillon de kératine prélevé chez des sujets présentant une pathologie
WO2008102557A1 (fr) 2007-02-21 2008-08-28 Oncotherapy Science, Inc. Vaccins peptidiques pour les cancers exprimant des antigènes associés à une tumeur
US20100040641A1 (en) 2007-02-21 2010-02-18 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing tumor-associated antigens
WO2009025116A1 (fr) 2007-08-20 2009-02-26 Oncotherapy Science, Inc. Peptide cdh3 et agent médicinal le comprenant

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
Baselga, J., "Herceptin® Alone or in Combination with Chemotherapy in the Treatment of HER2-Positive Metastatic Breast Cancer: Pivotal Trials," Oncology, vol. 61, Suppl. 2, pp. 14-21 (2001).
Behrens, J., et al., "Cadherins and catenins: Role in signal transduction and tumor progression," Cancer and Metastasis Reviews, vol. (1). 18, pp. 15-30 (1999).
Conacci-Sorrell, M., et al., "The cadherin-catenin adhesion system in signaling and cancer," The Journal of Clinical Investigation, vol. 109(8), pp. 987-991 (Apr. 2002).
Crist, W., et al., "Intergroup Rhabdomyosarcoma Study-IV: Results for Patients With Nonmetastatic Disease," Journal of Clinical Oncology, vol. 19(12), pp. 3091-3102 (Jun. 2001).
Daniel, C., et al., "Expression and Functional Role of E-and P-Cadherins in Mouse Mammary Ductal Morphogenesis and Growth," Dev. Biol., vol. 169(2), pp. 511-519 (Jun. 1995).
Ferguson, W., et al., "Current Treatment of Osteosarcoma," Cancer Investigation, vol. 19(3), pp. 292-315 (2001).
Ferrara, N., et al., "Discovery and Development of Bevacizumab, an Anti-VEGF Antibody for Treatment Cancer," Nat Rev Drug Discov., vol. 3(5), pp. 391-400 (May 2004).
Gamallo, C., et al., "The Prognostic Significance of P-Cadherin in Infiltrating Ductal Breast Carcinoma," Mod Pathol., vol. 14(7), pp. 650-654 (Jul. 2001).
Harris, M., "Monoclonal antibodies as therapeutic agents for cancer," Lancet Oncol., vol. 5(5), pp. 292-302 (May 2004).
Maloney, D., et al., "IDEC-C2B8 (Rituximab) Anti-CD20 Monoclonal Antibody Therapy in Patients With Relapsed Low-Grade Non-Hodgkin's Lymphoma," Blood, vol. 90(6), pp. 2188-2195 (Sep. 15, 1997).
Mialhe, et al., "Expression of E-, P-, N-Cadherins and Catenins in Human Bladder Carcinoma Cell Lines," J Urol., vol. 164(3 Pt 1), pp. 826-835 (Sep. 2000).
Nakamura, T., et al., "Genome-wide cDNA microarray analysis of gene expression profiles in pancreatic cancers using populations of tumor cells and normal ductal epithelial cells selected for purity by laser microdissection," Oncogene, vol. 23(13), pp. 2385-2400 (Mar. 25, 2004).
Nose, A., et al. "A Novel Cadherin Cell Adhesion Molecule: Its Expression Patterns Associated with Implantation and Organogenesis of Mouse Embryos," J Cell Biol., vol. 103(6, Pt. 2), pp. 2649-2658 (Dec. 1986).
Nose, A., et al., "Expressed Recombinant Cadherins Mediate Cell Sorting in Model Systems," Cell, vol. 54(7), pp. 993-1001 (Sep. 23, 1988).
Pyo, et al., "Expression of E-cadherin, P-cadherin and N-cadherin in oral squamous cell carcinoma: Correlation with the clinicopathologic features and patient outcome," J Craniomaxillofac Surg., vol. 35(1), pp. 1-9 (Epub Feb. 12, 2007, Jan. 2007).
Shimoyama, Y., et al., "Cadherin Cell-Adhesion Molecules in Human Epithelial Tissues and Carcinomas," Cancer Res., vol. 49(8), pp. 2128-2133 (Apr. 15, 1989).
Stefansson, I., et al., Prognostic Impact of Alterations in P-Cadherin Expression and Related Cell Adhesion Markers in Endometrial Cancer, J Clin Oncol., vol. 22(7), pp. 1242-1252 (Apr. 1, 2004).
Steinberg, M., et al., "Experimental specification of cell sorting, tissue spreading, and specific spatial patterning by quantitative differences in cadherin expression," Proc Natl Acad Sci USA. vol. 91(1), pp. 206-209 (Jan. 4, 1994).
Supplementary European Search Report for European Application EP 09773163, dated Dec. 7, 2012, 4 pages.
Takeichi, M., "Cadherin Cell Adhesion Receptors as a Morphogenetic Regulator," Science, vol. 251(5000), pp. 1451-1455 (Mar. 22, 1991).
Takeichi, M., "The cadherins: cell-cell adhesion molecules controlling animal morphogenesis," Development, vol. 102(4), pp. 639-655 (Apr. 1988).
U.S. Appl. No. 12/673,451, filed Jun. 5, 2008, 134 pgs.
U.S. Appl. No. 13/464,831, filed May 4, 2012, 163 pages.
U.S. Appl. No. 13/519,127, which is a U.S. National Stage of PCT/JP2009/007333, 59 pages, filed Dec. 28, 2009.
U.S. Appl. No. 13/536,327, 204 pages, filed Jun. 28, 2012.
U.S. Appl. No. 13/744,354, filed Jan. 17, 2013, 124 pages.
Vajdos et al. Comprehensive functional maps of the antigen-binding site of an anti-ErbB2 antibody obtained with shotgun scanning mutagenesis. J. Mol. Biol. Jul. 5, 2002, 320(2):415-28. *
Wu, et al., "JNK signaling pathway is required for bFGF-mediated surface cadherin downregulation on HUVEC," Exp Cell Res., vol. 314(3), pp. 421-429, doi: 10. 1016/j.yexcr (Epub Oct. 6, 2007, Feb. 1, 2008).
Wunder, J., et al., "The Histological Response to Chemotherapy as a Predictor of the Oncological Outcome of Operative Treatment of Ewing Sarcoma," J Bone Joint Surg Am, vol. 80(7), pp. 1020-1033 (Jul. 1998).

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130034566A1 (en) * 2009-12-28 2013-02-07 Onco Therapy Science, Inc. Anti-cdh3 antibodies and uses thereof
US9017669B2 (en) * 2009-12-28 2015-04-28 Oncotherapy Science, Inc. Anti-CDH3 antibodies and uses thereof
US20160058883A1 (en) * 2012-04-04 2016-03-03 Perseus Proteomics Inc. Drug conjugate comprising anti-cdh3 (p-cadherin) antibody
US9764041B2 (en) * 2012-04-04 2017-09-19 Perseus Proteomics Inc. Drug conjugate comprising anti-CDH3 (P-cadherin) antibody
US9884921B2 (en) 2014-07-01 2018-02-06 Pfizer Inc. Bispecific heterodimeric diabodies and uses thereof
US20180193477A1 (en) * 2015-07-15 2018-07-12 Zymeworks Inc. Drug-conjugated bi-specific antigen-binding constructs
US11147886B2 (en) * 2015-07-15 2021-10-19 Zymeworks Inc. Drug-conjugated bi-specific antigen-binding constructs

Also Published As

Publication number Publication date
RU2011103199A (ru) 2012-08-10
CA2729403A1 (fr) 2010-01-07
CN102137929A (zh) 2011-07-27
CN102137929B (zh) 2013-07-10
JP5593560B2 (ja) 2014-09-24
WO2010001585A1 (fr) 2010-01-07
EP2313504A1 (fr) 2011-04-27
US20120128584A1 (en) 2012-05-24
ES2534437T3 (es) 2015-04-22
KR20110025215A (ko) 2011-03-09
JP2011526583A (ja) 2011-10-13
BRPI0913656A2 (pt) 2019-09-24
EP2313504B1 (fr) 2015-01-07
EP2313504A4 (fr) 2013-01-23

Similar Documents

Publication Publication Date Title
US8435749B2 (en) Anti-CDH3 antibodies labeled with radioisotope label and uses thereof
US9017669B2 (en) Anti-CDH3 antibodies and uses thereof
ES2405618T3 (es) Anticuerpos monoclonales dirigidos a tumores para FZD10 y usos de los mismos
KR20100003366A (ko) 암 세포의 세포독성을 매개하는 인간화 및 키메라 항-trop-2 항체
JP2011526583A5 (fr)
JP2010530361A (ja) Cd59の表面発現を証明する細胞の細胞毒性媒介
US10407503B2 (en) Fully human antibodies and fragments recognizing human c-Met
BRPI0807446A2 (pt) Anticorpos modificantes de doenças cancerosa
AU2007278791A1 (en) Cytotoxicity mediation of cells evidencing surface expression of CD63
TW200922619A (en) Cancerous disease modifying antibodies
JP2009528993A (ja) 癌様疾患修飾性抗体141205−02
BRPI0718644A2 (pt) Anticorpo monoclonal isolado, anticorpos humanizado e quimérico do anticorpo monoclonal isolado, linha de célula hibridoma isolada, método para iniciar a citotoxidez induzida por anticorpo de células cancerosas em uma amostra de tecido selecionada de um tumor humano, cdmab do anticorpo monoclonal isolado ou seu cdmab e usos de um anticorpo monoclonal ou seu cdmab, sozinho ou em conjução com pelo menos um agente quimioterapêutico, e composição
CN105111313B (zh) 抗cdh3抗体及其用途
JP2010523479A (ja) ハイブリドーマ細胞系列ar51a630.3により産生される癌性疾患修飾性抗体010207−01
US20080241062A1 (en) Cancerous disease modifying antibodies
NZ626513B2 (en) Anti-cd98 antibodies and methods of use thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: ONCOTHERAPY SCIENCE, INC., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:TOGASHI, AKIRA;KATSU, MASAKAZU;TAKAYANAGI, MEGUMI;AND OTHERS;SIGNING DATES FROM 20101008 TO 20101202;REEL/FRAME:026058/0605

Owner name: ONCOTHERAPY SCIENCE, INC., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THE UNIVERSITY OF TOKYO;REEL/FRAME:026058/0655

Effective date: 20110117

Owner name: NATIONAL UNIVERSITY CORPORATION GUNMA UNIVERSITY,

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ENDO, KEIGO;REEL/FRAME:026058/0631

Effective date: 20101108

Owner name: ONCOTHERAPY SCIENCE, INC., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NATIONAL UNIVERSITY CORPORATION GUNMA UNIVERSITY;REEL/FRAME:026058/0648

Effective date: 20101222

Owner name: ONCOTHERAPY SCIENCE, INC., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NOSAN CORPORATION;REEL/FRAME:026058/0644

Effective date: 20101025

Owner name: NOSAN CORPORATION, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NG, POHSING;REEL/FRAME:026058/0619

Effective date: 20101021

Owner name: THE UNIVERSITY OF TOKYO, JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NAKAMURA, YUSUKE;REEL/FRAME:026058/0641

Effective date: 20101122

STCF Information on status: patent grant

Free format text: PATENTED CASE

CC Certificate of correction
FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FEPP Fee payment procedure

Free format text: PAT HOLDER CLAIMS SMALL ENTITY STATUS, ENTITY STATUS SET TO SMALL (ORIGINAL EVENT CODE: LTOS); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YR, SMALL ENTITY (ORIGINAL EVENT CODE: M2552); ENTITY STATUS OF PATENT OWNER: SMALL ENTITY

Year of fee payment: 8